Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Biomolecules ; 14(2)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38397400

ABSTRACT

Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.


Subject(s)
Depression , Migraine Disorders , Humans , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Migraine Disorders/drug therapy , Migraine Disorders/etiology , Calcitonin Gene-Related Peptide , Neuropeptide Y
2.
Brain ; 147(4): 1312-1320, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-37864847

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide-38 (PACAP-38), known for its role in migraine pathogenesis, has been identified as a novel drug target. Given the clinical parallels between post-traumatic headache (PTH) and migraine, we explored the possible role of PACAP-38 in the pathogenesis of PTH. To this end, we conducted a randomized, double-blind, placebo-controlled, two-way crossover trial involving adult participants diagnosed with persistent PTH resulting from mild traumatic brain injury. Participants were randomly assigned to receive a 20-min continuous intravenous infusion of either PACAP-38 (10 pmol/kg/min) or placebo (isotonic saline) on two separate experimental days, with a 1-week washout period in between. The primary outcome was the difference in incidence of migraine-like headache between PACAP-38 and placebo during a 12-h observational period post-infusion. The secondary outcome was the difference in the area under the curve (AUC) for baseline-corrected median headache intensity scores during the same 12-h observational period. Of 49 individuals assessed for eligibility, 21 were enrolled and completed the trial. The participants had a mean age of 35.2 years, and 16 (76%) were female. Most [19 of 21 (90%)] had a migraine-like phenotype. During the 12-h observational period, 20 of 21 (95%) participants developed migraine-like headache after intravenous infusion of PACAP-38, compared with two (10%) participants after placebo (P < 0.001). Furthermore, the baseline-corrected AUC values for median headache intensity scores during the 12-h observational period was higher after PACAP-38 than placebo (P < 0.001). These compelling results demonstrate that PACAP-38 is potent inducer of migraine-like headache in people with persistent PTH. Thus, targeting PACAP-38 signalling might be a promising avenue for the treatment of PTH.


Subject(s)
Migraine Disorders , Post-Traumatic Headache , Adult , Humans , Female , Male , Post-Traumatic Headache/drug therapy , Post-Traumatic Headache/diagnosis , Post-Traumatic Headache/etiology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Headache/etiology , Headache/complications , Migraine Disorders/drug therapy , Migraine Disorders/complications , Double-Blind Method
3.
Int J Mol Sci ; 24(17)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37686074

ABSTRACT

Despite the high probability of glaucoma-related blindness, its cause is not fully understood and there is no efficient therapeutic strategy for neuroprotection. Vascular factors have been suggested to play an important role in glaucoma development and progression. Previously, we have proven the neuroprotective effects of pituitary adenylate-cyclase-activating polypeptide (PACAP) eye drops in an inducible, microbeads model in rats that is able to reproduce many clinically relevant features of human glaucoma. In the present study, we examined the potential protective effects of PACAP1-38 on the retinal vasculature and the molecular changes in hypoxia. Ocular hypertension was induced by injection of microbeads into the anterior chamber, while control rats received PBS. PACAP dissolved in vehicle (1 µg/drop) or vehicle treatment was started one day after the injections for four weeks three times a day. Retinal degeneration was assessed with optical coherence tomography (OCT), and vascular and molecular changes were assessed by immunofluorescence labeling. HIF1-α and VEGF-A protein levels were measured by Western blot. OCT images proved severe retinal degeneration in the glaucomatous group, while PACAP1-38 eye drops had a retinoprotective effect. Vascular parameters were deteriorated and molecular analysis suggested hypoxic conditions in glaucoma. PACAP treatment exerted a positive effect against these alterations. In summary, PACAP could prevent the severe damage to the retina and its vasculature induced by ocular hypertension in a microbeads model.


Subject(s)
Glaucoma , Ocular Hypertension , Retinal Degeneration , Animals , Rats , Glaucoma/drug therapy , Hypoxia , Ocular Hypertension/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Retinal Vessels
4.
Int J Mol Sci ; 24(15)2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37569369

ABSTRACT

Migraine is a common condition with disabling attacks that burdens people in the prime of their working lives. Despite years of research into migraine pathophysiology and therapeutics, much remains to be learned about the mechanisms at play in this complex neurovascular condition. Additionally, there remains a relative paucity of specific and targeted therapies available. Many sufferers remain underserved by currently available broad action preventive strategies, which are also complicated by poor tolerance and adverse effects. The development of preclinical migraine models in the laboratory, and the advances in human experimental migraine provocation, have led to the identification of key molecules likely involved in the molecular circuity of migraine, and have provided novel therapeutic targets. Importantly, the identification that vasoconstriction is neither necessary nor required for headache abortion has changed the landscape of migraine treatment and has broadened the therapy targets for patients with vascular risk factors or vascular disease. These targets include nitric oxide synthase (NOS) and several neuropeptides that are involved in migraine. The ability of NO donors and infusion of some of these peptides into humans to trigger typical migraine-like attacks has supported the development of targeted therapies against these molecules. Some of these, such as those targeting calcitonin gene-related peptide (CGRP), have already reached clinical practice and are displaying a positive outcome in migraineurs for the better by offering targeted efficacy without significant adverse effects. Others, such as those targeting pituitary adenylate cyclase activating polypeptide (PACAP), are showing promise and are likely to enter phase 3 clinical trials in the near future. Understanding these nitrergic and peptidergic mechanisms in migraine and their interactions is likely to lead to further therapeutic strategies for migraine in the future.


Subject(s)
Migraine Disorders , Humans , Migraine Disorders/drug therapy , Headache , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Calcitonin Gene-Related Peptide , Nitric Oxide Synthase
5.
Curr Neurovasc Res ; 20(3): 296-313, 2023.
Article in English | MEDLINE | ID: mdl-37488760

ABSTRACT

BACKGROUND: There has been a protracted effort to identify reliable targets for migraine. It is believed that each year, hundreds of millions of individuals worldwide suffer from migraines, making this widespread neurological ailment the second leading cause of years of disability worldwide. The rationale of this study is to identify the major targets involved in migraine attacks. METHODS: For this review, specialized databases were searched, such as PubMed, EMBASE, DynaMed Plus, and Science Direct databases that included the pathophysiological mechanisms of migraine, focusing on in vitro and in vivo studies in the clinical management of migraine. RESULTS: Calcitonin gene-related peptide, Pituitary adenylate cyclase-activating polypeptide (PACAP), NOD-like receptor Protein (NLRP3), Serotonin, and some other neuroinflammatory biomarkers are collectively responsible for the cerebral blood vessel dilation and involved in the nociceptive pain which leads to migraine attack. CONCLUSION: Migraine biomarkers such as CGRP, PACAP, NLRP3, Nitric oxide synthase, MMP9, and Serotonin could be targets for developing drugs. Present marketed medications temporarily reduce symptoms and pain and have serious cardiovascular side effects. It is suggested that herbal treatment may help prevent migraine attacks without adverse effects. Natural biomolecules that may give better treatment than the present marketed medication and full fledge research should be carried out with natural biomarkers by the Network Pharmacological approach.


Subject(s)
Migraine Disorders , Pituitary Adenylate Cyclase-Activating Polypeptide , Humans , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Serotonin , Migraine Disorders/drug therapy , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/therapeutic use
6.
J Neuroinflammation ; 19(1): 274, 2022 Nov 19.
Article in English | MEDLINE | ID: mdl-36403002

ABSTRACT

BACKGROUND: Cerebral infection with the protozoan Toxoplasma gondii (T. gondii) is responsible for inflammation of the central nervous system (CNS) contributing to subtle neuronal alterations. Albeit essential for brain parasite control, continuous microglia activation and recruitment of peripheral immune cells entail distinct neuronal impairment upon infection-induced neuroinflammation. PACAP is an endogenous neuropeptide known to inhibit inflammation and promote neuronal survival. Since PACAP is actively transported into the CNS, we aimed to assess the impact of PACAP on the T. gondii-induced neuroinflammation and subsequent effects on neuronal homeostasis. METHODS: Exogenous PACAP was administered intraperitoneally in the chronic stage of T. gondii infection, and brains were isolated for histopathological analysis and determination of pathogen levels. Immune cells from the brain, blood, and spleen were analyzed by flow cytometry, and the further production of inflammatory mediators was investigated by intracellular protein staining as well as expression levels by RT-qPCR. Neuronal and synaptic alterations were assessed on the transcriptional and protein level, focusing on neurotrophins, neurotrophin-receptors and signature synaptic markers. RESULTS: Here, we reveal that PACAP administration reduced the inflammatory foci and the number of apoptotic cells in the brain parenchyma and restrained the activation of microglia and recruitment of monocytes. The neuropeptide reduced the expression of inflammatory mediators such as IFN-γ, IL-6, iNOS, and IL-1ß. Moreover, PACAP diminished IFN-γ production by recruited CD4+ T cells in the CNS. Importantly, PACAP promoted neuronal health via increased expression of the neurotrophin BDNF and reduction of p75NTR, a receptor related to neuronal cell death. In addition, PACAP administration was associated with increased expression of transporters involved in glutamatergic and GABAergic signaling that are particularly affected during cerebral toxoplasmosis. CONCLUSIONS: Together, our findings unravel the beneficial effects of exogenous PACAP treatment upon infection-induced neuroinflammation, highlighting the potential implication of neuropeptides to promote neuronal survival and minimize synaptic prejudice.


Subject(s)
Toxoplasma , Toxoplasmosis , Humans , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Neuroinflammatory Diseases , Toxoplasmosis/complications , Toxoplasmosis/drug therapy , Nerve Growth Factors , Inflammation/drug therapy , Inflammation Mediators
7.
Biochem Biophys Res Commun ; 631: 146-151, 2022 11 26.
Article in English | MEDLINE | ID: mdl-36194909

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a highly conserved pleiotropic neuropeptide, implicated in emotional stress responses and anxiety-related disorders. Here, we examined whether our recently developed small-molecule non-peptide PACAP receptor antagonists could ameliorate anxiety-like behaviors induced by acute restraint stress in mice. The antagonists PA-9 and its derivative PA-915 improved anxiety-like behaviors in mice subjected to restraint stress. An anxiolytic effect was observed with single acute dose, suggesting their fast-acting properties. PA-915 demonstrated a statistically significant anxiolytic effect whereas fluoxetine did not. These results indicate the potential of PAC1 antagonists as a novel treatment for anxiety.


Subject(s)
Anti-Anxiety Agents , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Fluoxetine , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
8.
Expert Opin Pharmacother ; 23(11): 1325-1335, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35850597

ABSTRACT

INTRODUCTION: Migraine occupies the first position regarding the disability caused in female working population (15-49 years). Research in the field of prophylaxis in this pathology has made enormous strides in recent years. AREAS COVERED: In this narrative review, we retrace the most important scientific evidence regarding recently approved and emerging drugs for the prophylactic treatment of migraine. The purpose of this article is in fact to evaluate currently approved or emerging pharmacological agents for migraine prophylaxis. This review is based on the literature published in the peer review journal obtained through PubMed, Cochrane library, Clinicaltrials.gov, and US FDA. EXPERT OPINION: Monoclonal antibodies (mAbs) that target the calcitonin gene-related peptide signaling pathway (CGRP) have marked an innovation in prophylactic migraine therapy. The combination of Onabotulinumtoxin-A (OBTA) and mAbs appears to be an effective, but costly, therapeutic option for resistant cases. New classes of molecules like gepants and ditans seem to give exceptional results. In addition, new prophylactic drugs are emerging with several targets: the pituitary adenylate cyclase-activating polypeptide (PACAP), ion channels, several receptors coupled to G proteins, orexin, and glutamate. All these therapies will implement and improve migraine management, as well as personalized medicine for each patient.


Subject(s)
Calcitonin Gene-Related Peptide Receptor Antagonists , Migraine Disorders , Antibodies, Monoclonal/therapeutic use , Calcitonin Gene-Related Peptide/therapeutic use , Calcitonin Gene-Related Peptide Receptor Antagonists/therapeutic use , Humans , Migraine Disorders/drug therapy , Migraine Disorders/prevention & control , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use
9.
Curr Opin Neurol ; 35(3): 336-342, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35674077

ABSTRACT

PURPOSE OF REVIEW: The pathophysiological understanding of cluster headache has evolved significantly over the past years. Although it is now well known that the trigeminovascular system, the parasympathetic system and the hypothalamus play important roles in its pathomechanism, we increasingly understand the functional role several neurotransmitters and hormones play in the communication between these structures. RECENT FINDINGS: This work will give an overview of the current understanding of the role of calcitonin gene-related peptide, vasoactive intestinal peptide, pituitary adenylate cyclase-activating peptide, melatonin and orexins in cluster headache. On the basis of recent evidence, this study will also review the relevance of the monoclonal calcitonin gene-related peptide antibody galcanezumab as well as the sleep-regulating hormone melatonin in the treatment of cluster headache. SUMMARY: Herein, we aim to review the basic mechanisms implicated in the pathophysiology of cluster headache and how the increased mechanistic understanding may lead to the discovery of novel therapeutic targets.


Subject(s)
Cluster Headache , Melatonin , Antibodies, Monoclonal/therapeutic use , Calcitonin Gene-Related Peptide , Cluster Headache/drug therapy , Humans , Melatonin/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Vasoactive Intestinal Peptide/therapeutic use
10.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054857

ABSTRACT

Dry eye disease (DED) is caused by a reduction in the volume or quality of tears. The prevalence of DED is estimated to be 100 million in the developed world. As aging is a risk factor for DED, the prevalence of DED is expected to grow at a rapid pace in aging populations, thus creating an increased need for new therapies. This review summarizes DED medications currently in clinical use. Most current medications for DED focus on stimulating tear secretion, mucin secretion, or suppressing inflammation, rather than simply replenishing the ocular surface with moisture to improve symptoms. We recently reported that the neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) induces tear secretion and suppresses corneal injury caused by a reduction in tears. Moreover, it has been reported that a PACAP in water and a 0.9% saline solution at +4 °C showed high stability and achieved 80-90% effectiveness after 2 weeks of treatment. These results reveal PACAP as a candidate DED medication. Further research on the clinical applications of PACAP in DED is necessary.


Subject(s)
Dry Eye Syndromes/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Animals , Dry Eye Syndromes/pathology , Humans , Models, Biological , Ophthalmic Solutions/pharmacology , Ophthalmic Solutions/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Signal Transduction/drug effects , Tears/drug effects
11.
Curr Opin Endocrinol Diabetes Obes ; 29(2): 225-231, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35066541

ABSTRACT

PURPOSE OF REVIEW: To review the latest advances in migraine management with a focus on medications specifically developed for the treatment of migraine. RECENT FINDINGS: Randomized clinical trials demonstrated the efficacy of calcitonin gene-related peptide (CGRP) mAbs for the preventive treatment of migraine and the small molecule CGRP receptor antagonist gepants for acute abortion and prevention of migraine attacks. Pituitary adenylate cyclase-activating polypeptide (PACAP) is another signaling molecule of interest and represents a potential new drug class of mechanism-based migraine medications. Drugs targeting PACAP are currently undergoing clinical trials, and the coming years will reveal whether this class of drugs will expand our therapeutic armamentarium. SUMMARY: Here, we summarize the role of CGRP and PACAP in migraine pathophysiology and discuss novel therapies targeting the CGRP and PACAP signaling pathways.


Subject(s)
Calcitonin Gene-Related Peptide , Migraine Disorders , Antibodies, Monoclonal/therapeutic use , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/therapeutic use , Humans , Migraine Disorders/drug therapy , Migraine Disorders/prevention & control , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Signal Transduction
12.
Peptides ; 146: 170630, 2021 12.
Article in English | MEDLINE | ID: mdl-34481915

ABSTRACT

Prenatal ethanol exposure provokes teratogenic effects, due to oxidative stress and massive neuronal apoptosis in the developing brain that result in lifelong behavioral abnormalities. PACAP exerts anti-oxidative and neuroprotective activities on neuronal cells, and prevents ethanol neurotoxicity. The present study focused on the ability of PACAP to protect the brain of 30-day-old mice (P30) from prenatal alcohol exposure induced oxidative damage and toxicity. Pregnant mice were divided randomly into 4 groups, i.e. control group, ethanol group (1.5 g/kg ip daily injection), PACAP group (5 µg intrauterine daily injection) and an ethanol plus PACAP group. Offspring prenatally exposed to ethanol had decreased body weight and reduced cell survival. Moreover, production of ROS was sharply enhanced in the brain of prenatal ethanol-exposed animals, associated with an elevation in the activity of the antioxidant enzymes, and an increase of oxidative damages as shown by the accumulation of the lipid oxidation marker malondialdehyde and of protein carbonyl compounds. Intrauterine administration of PACAP during the gestational period restored the endogenous antioxidant system, prevented ROS overproduction and promoted the survival of dissociated cells from animals prenatally exposed to ethanol. Behavioral tests revealed that P30 animals exposed to ethanol during the prenatal period exhibited reduced motor activity, altered exploratory interest and increased anxiety. However, PACAP treatment significantly attenuated these behavioral impairments. This study demonstrates that PACAP exerts a potent neuroprotective effect against alcohol toxicity during brain development, and indicates that PACAP and/or PACAP analogs might be a useful tool for treatment of alcohol intoxication during pregnancy.


Subject(s)
Fetal Alcohol Spectrum Disorders/prevention & control , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Cell Survival/drug effects , Cognition Disorders/prevention & control , Disease Models, Animal , Down-Regulation/drug effects , Female , Mice, Inbred C57BL , Movement Disorders/prevention & control , Oxidative Stress/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects
13.
Pain ; 162(5): 1483-1499, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33252452

ABSTRACT

A large body of animal and human studies indicates that blocking peripheral calcitonin gene-related peptide (CGRP) and pituitary adenylate cyclase-activating polypeptide (PACAP) signaling pathways may prevent migraine episodes and reduce headache frequency. To investigate whether recurring migraine episodes alter the strength of CGRP and PACAP signaling in trigeminal ganglion (TG) neurons, we compared the number of TG neurons that respond to CGRP and to PACAP (CGRP-R and PACAP-R, respectively) under normal and chronic migraine-like conditions. In a mouse model of chronic migraine, repeated nitroglycerin (NTG) administration significantly increased the number of CGRP-R and PACAP-R neurons in TG but not dorsal root ganglia. In TG neurons that express endogenous αCGRP, repeated NTG led to a 7-fold increase in the number of neurons that respond to both CGRP and PACAP (CGRP-R&PACAP-R). Most of these neurons were unmyelinated C-fiber nociceptors. This suggests that a larger fraction of CGRP signaling in TG nociceptors may be mediated through the autocrine mechanism, and the release of endogenous αCGRP can be enhanced by both CGRP and PACAP signaling pathways under chronic migraine condition. The number of CGRP-R&PACAP-R TG neurons was also increased in a mouse model of posttraumatic headache (PTH). Interestingly, low-dose interleukin-2 treatment, which completely reverses chronic migraine-related and PTH-related behaviors in mouse models, also blocked the increase in both CGRP-R and PACAP-R TG neurons. Together, these results suggest that inhibition of both CGRP and PACAP signaling in TG neurons may be more effective in treating chronic migraine and PTH than targeting individual signaling pathways.


Subject(s)
Migraine Disorders , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Calcitonin Gene-Related Peptide , Headache , Mice , Migraine Disorders/drug therapy , Neurons , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Trigeminal Ganglion
14.
Int J Mol Sci ; 21(21)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153152

ABSTRACT

Schwann cells, the most abundant glial cells of the peripheral nervous system, represent the key players able to supply extracellular microenvironment for axonal regrowth and restoration of myelin sheaths on regenerating axons. Following nerve injury, Schwann cells respond adaptively to damage by acquiring a new phenotype. In particular, some of them localize in the distal stump to form the Bungner band, a regeneration track in the distal site of the injured nerve, whereas others produce cytokines involved in recruitment of macrophages infiltrating into the nerve damaged area for axonal and myelin debris clearance. Several neurotrophic factors, including pituitary adenylyl cyclase-activating peptide (PACAP), promote survival and axonal elongation of injured neurons. The present review summarizes the evidence existing in the literature demonstrating the autocrine and/or paracrine action exerted by PACAP to promote remyelination and ameliorate the peripheral nerve inflammatory response following nerve injury.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Schwann Cells/drug effects , Trauma, Nervous System , Animals , Axons/drug effects , Axons/physiology , Cell Survival/drug effects , Humans , Myelin Sheath/drug effects , Myelin Sheath/physiology , Nerve Fibers/drug effects , Nerve Fibers/physiology , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Peripheral Nerves/drug effects , Peripheral Nerves/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Schwann Cells/physiology , Trauma, Nervous System/drug therapy , Trauma, Nervous System/pathology , Trauma, Nervous System/physiopathology
15.
Histol Histopathol ; 35(11): 1251-1262, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32542641

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is an evolutionally well conserved neuropeptide, mainly expressed by neuronal and peripheral cells. It proves to be an interesting object of study both for its trophic functions during the development of several tissues and for its protective effects against oxidative stress, hypoxia, inflammation and apoptosis in different degenerative diseases. This brief review summarises the recent findings concerning the role of PACAP in the articular cartilage. PACAP and its receptors are expressed during chondrogenesis and are shown to activate the pathways involved in regulating cartilage development. Moreover, this neuropeptide proves to be chondroprotective against those stressors that determine cartilage degeneration and contribute to the onset of osteoarthritis (OA), the most common form of degenerative joint disease. Indeed, the degenerated cartilage exhibits low levels of PACAP, suggesting that its endogenous levels in adult cartilage may play an essential role in maintaining physiological properties. Thanks to its peculiar characteristics, exogenous administration of PACAP could be suggested as a potential tool to slow down the progression of OA and for cartilage regeneration approaches.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrogenesis , Osteoarthritis/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Antirheumatic Agents/therapeutic use , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Chondrocytes/drug effects , Chondrocytes/pathology , Chondrogenesis/drug effects , Humans , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction
16.
Theranostics ; 10(10): 4453-4465, 2020.
Article in English | MEDLINE | ID: mdl-32292507

ABSTRACT

Organ ischemia reperfusion injury (IRI), associated with acute hepatocyte death, remains an unresolved problem in clinical orthotopic liver transplantation (OLT). Autophagy, an intracellular self-digesting progress, is responsible for cell reprograming required to regain post-stress homeostasis. Methods: Here, we analyzed the cytoprotective mechanism of pituitary adenylate cyclase-activating polypeptide (PACAP)-promoted hepatocellular autophagy in a clinically relevant mouse model of extended hepatic cold storage (4 °C UW solution for 20 h) followed by syngeneic OLT. Results: In contrast to 41.7% of liver graft failure by day 7 post-transplant in control group, PACAP treatment significantly improved graft survival (91.7% by day 14), and promoted autophagy-associated regeneration programs in OLT. In parallel in vitro studies, PACAP-enhanced autophagy ameliorated cellular damage (LDH/ALT levels), and diminished necrosis in H2O2-stressed primary hepatocytes. Interestingly, PACAP not only induced nuclear cAMP response element-binding protein (CREB), but also triggered reprogramming factor Kruppel-like factor 4 (KLF4) expression in IR-stressed OLT. Indeed, CREB inhibition attenuated hepatic autophagy and recreated hepatocellular injury in otherwise PACAP-protected livers. Furthermore, CREB inhibition suppressed PACAP-induced KLF4 expression, whereas KLF4 blockade abolished PACAP-promoted autophagy and neutralized PACAP-mediated hepatoprotection both in vivo and in vitro. Conclusion: Current study documents the essential neural regulation of PACAP-promoted autophagy in hepatocellular homeostasis in OLT, which provides the emerging therapeutic principle to combat hepatic IRI in OLT.


Subject(s)
Autophagy/drug effects , Hepatocytes/cytology , Liver/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Primary Graft Dysfunction/drug therapy , Reperfusion Injury , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Hepatocytes/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Liver/cytology , Liver Transplantation , Male , Mice , Mice, Inbred C57BL , Specimen Handling
17.
Pain ; 161(7): 1670-1681, 2020 07.
Article in English | MEDLINE | ID: mdl-32142016

ABSTRACT

Pituitary adenylate cyclase activating polypeptide-38 (PACAP38) may play an important role in primary headaches. Preclinical evidence suggests that PACAP38 modulates trigeminal nociceptive activity mainly through PAC1 receptors while clinical studies report that plasma concentrations of PACAP38 are elevated in spontaneous attacks of cluster headache and migraine and normalize after treatment with sumatriptan. Intravenous infusion of PACAP38 induces migraine-like attacks in migraineurs and cluster-like attacks in cluster headache patients. A rodent-specific PAC1 receptor antibody Ab181 was developed, and its effect on nociceptive neuronal activity in the trigeminocervical complex was investigated in vivo in an electrophysiological model relevant to primary headaches. Ab181 is potent and selective at the rat PAC1 receptor and provides near-maximum target coverage at 10 mg/kg for more than 48 hours. Without affecting spontaneous neuronal activity, Ab181 effectively inhibits stimulus-evoked activity in the trigeminocervical complex. Immunohistochemical analysis revealed its binding in the trigeminal ganglion and sphenopalatine ganglion but not within the central nervous system suggesting a peripheral site of action. The pharmacological approach using a specific PAC1 receptor antibody could provide a novel mechanism with a potential clinical efficacy in the treatment of primary headaches.


Subject(s)
Migraine Disorders , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Headache/chemically induced , Headache/drug therapy , Humans , Nociception , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Rats
18.
Brain Behav ; 10(1): e01465, 2020 01.
Article in English | MEDLINE | ID: mdl-31769222

ABSTRACT

BACKGROUND: Inflammation in the brain is mediated by the cyclooxygenase pathway, which leads to the production of prostaglandins. Prostaglandin (PG) D2, the most abundant PG in the brain, increases under pathological conditions and is spontaneously metabolized to PGJ2. PGJ2 is highly neurotoxic, with the potential to transition neuroinflammation into a chronic state and contribute to neurodegeneration as seen in many neurological diseases. Conversely, PACAP27 is a lipophilic peptide that raises intracellular cAMP and is an anti-inflammatory agent. The aim of our study was to investigate the therapeutic potential of PACAP27 to counter the behavioral and neurotoxic effects of PGJ2 observed in aged subjects. METHODS: PGJ2 was injected bilaterally into the hippocampal CA1 region of 53-week-old and 12-week-old C57BL/6N male mice, once per week over 3 weeks (three total infusions) and included co-infusions of PACAP27 within respective treatment groups. Our behavioral assessments looked at spatial learning and memory performance on the 8-arm radial maze, followed by histological analyses of fixed hippocampal tissue using Fluoro-Jade C and fluorescent immunohistochemistry focused on IBA-1 microglia. RESULTS: Aged mice treated with PGJ2 exhibited spatial learning and long-term memory deficits, as well as neurodegeneration in CA3 pyramidal neurons. Aged mice that received co-infusions of PACAP27 exhibited remediated learning and memory performance and decreased neurodegeneration in CA3 pyramidal neurons. Moreover, microglial activation in the CA3 region was also reduced in aged mice cotreated with PACAP27. CONCLUSIONS: Our data show that PGJ2 can produce a retrograde spread of damage not observed in PGJ2-treated young mice, leading to age-dependent neurodegeneration of hippocampal neurons producing learning and memory deficits. PACAP27 can remediate the behavioral and neurodegenerative effects that PGJ2 produces in aged subjects. Targeting specific neurotoxic prostaglandins, such as PGJ2, offers great promise as a new therapeutic strategy downstream of cyclooxygenases, to combat the neuronal deficits induced by chronic inflammation.


Subject(s)
Hippocampus/drug effects , Memory Disorders/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Prostaglandin D2/analogs & derivatives , Spatial Learning/drug effects , Animals , Hippocampus/metabolism , Male , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use
19.
J Mol Neurosci ; 68(3): 475-484, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30993644

ABSTRACT

Retinal ganglion cell (RGC) apoptosis is considered an important pathological hallmark of glaucoma. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic peptide with potent neuroprotective properties. In our previous study, we found that the expression of PACAP and its high-affinity receptor PACAP receptor type 1 (PAC1R) increased markedly after optic nerve crush (ONC), and occurred mainly in the ganglion cell layer of the retina. This suggests that the upregulation of PACAP may play a vital role in inhibiting RGC death after ONC. Therefore, in the present study, we investigate the specific effects and underlying mechanism of PACAP in RGC death after ONC. Vehicle (physiological saline) or PACAP (1 nM to 200 nM) solution was injected into the vitreous body. Seven days later, the retinas were harvested, and the surviving RGCs were retrogradely labeled with Fluoro-Gold (FG; Fluorochrome) at different concentrations of PACAP. Immunofluorescence double staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay were used to observe the effects of PACAP on RGC apoptosis. Our results showed that PACAP treatment inhibited caspase-3-mediated RGC apoptosis, promoted the phosphorylation of cAMP response element binding protein (CREB), up-regulated the expression of B-cell lymphoma 2 (Bcl-2), and ultimately improved RGC survival. These results suggest that PACAP may prevent RGC apoptosis after ONC via activation of CREB-mediated Bcl-2 transcription. The study thus contributes to a basic understanding of the mechanism by which PACAP decreased RGC apoptosis and provides a theoretical basis for future clinical application of PACAP in the treatment of glaucoma.


Subject(s)
Apoptosis , Neuroprotective Agents/pharmacology , Optic Atrophy/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Nerve Crush , Neuroprotective Agents/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Signal Transduction
20.
Neurochem Int ; 124: 238-244, 2019 03.
Article in English | MEDLINE | ID: mdl-30682380

ABSTRACT

Alcohol addiction is a worldwide concern as its detrimental effects go far beyond the addicted individual and can affect the entire family as well as the community. Considerable effort is being expended in understanding the neurobiological basis of such addiction in hope of developing effective prevention and/or intervention strategies. In addition, organ damage and neurotoxicological effects of alcohol are intensely investigated. Pharmacological approaches, so far, have only provided partial success in prevention or treatment of alcohol use disorder (AUD) including the neurotoxicological consequences of heavy drinking. Pituitary adenylate cyclase-activating polypeptide (PACAP) is an endogenous 38 amino-acid neuropeptide with demonstrated protection against neuronal injury, trauma as well as various endogenous and exogenous toxic agents including alcohol. In this mini-review, following a brief presentation of alcohol addiction and its neurotoxicity, the potential of PACAP as a therapeutic intervention in toxicological consequences of this devastating disorder is discussed.


Subject(s)
Alcoholic Intoxication/drug therapy , Brain/drug effects , Ethanol/toxicity , Neuroprotective Agents/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Alcoholic Intoxication/metabolism , Alcoholic Intoxication/pathology , Animals , Brain/metabolism , Brain/pathology , Ethanol/administration & dosage , Humans , Neuroprotective Agents/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...